Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.808
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 40(2): e105699, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33347626

RESUMEN

Pathogen type 3 secretion systems (T3SS) manipulate host cell pathways by directly delivering effector proteins into host cells. In Vibrio parahaemolyticus, the leading cause of bacterial seafood-borne diarrheal disease, we showed that a T3SS effector, VgpA, localizes to the host cell nucleolus where it binds Epstein-Barr virus nuclear antigen 1-binding protein 2 (EBP2). An amino acid substitution in VgpA (VgpAL10A ) did not alter its translocation to the nucleus but abolished the effector's capacity to interact with EBP2. VgpA-EBP2 interaction led to the re-localization of c-Myc to the nucleolus and increased cellular rRNA expression and proliferation of cultured cells. The VgpA-EBP2 interaction elevated EBP2's affinity for c-Myc and prolonged the oncoprotein's half-life. Studies in infant rabbits demonstrated that VgpA is translocated into intestinal epithelial cells, where it interacts with EBP2 and leads to nucleolar re-localization of c-Myc. Moreover, the in vivo VgpA-EBP2 interaction during infection led to proliferation of intestinal cells and heightened V. parahaemolyticus' colonization and virulence. These observations suggest that direct effector stimulation of a c-Myc controlled host cell growth program can contribute to pathogenesis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Nucléolo Celular/metabolismo , Proliferación Celular/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Vibrio parahaemolyticus/metabolismo , Virulencia/fisiología , Animales , Células CACO-2 , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células Epiteliales/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Herpesvirus Humano 4/patogenicidad , Humanos , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/metabolismo , Conejos , Vibriosis/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35131853

RESUMEN

Bacterial cells interact with solid surfaces and change their lifestyle from single free-swimming cells to sessile communal structures (biofilms). Cyclic di-guanosine monophosphate (c-di-GMP) is central to this process, yet we lack tools for direct dynamic visualization of c-di-GMP in single cells. Here, we developed a fluorescent protein-based c-di-GMP-sensing system for Escherichia coli that allowed us to visualize initial signaling events and assess the role played by the flagellar motor. The sensor was pH sensitive, and the events that appeared on a seconds' timescale were alkaline spikes in the intracellular pH. These spikes were not apparent when signals from different cells were averaged. Instead, a signal appeared on a minutes' timescale that proved to be due to an increase in intracellular c-di-GMP. This increase, but not the alkaline spikes, depended upon a functional flagellar motor. The kinetics and the amplitude of both the pH and c-di-GMP responses displayed cell-to-cell variability indicative of the distinct ways the cells approached and interacted with the surface. The energetic status of a cell can modulate these events. In particular, the alkaline spikes displayed an oscillatory behavior and the c-di-GMP increase was modest in the presence of glucose.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Transducción de Señal/fisiología , GMP Cíclico/metabolismo , Flagelos/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Vidrio , Concentración de Iones de Hidrógeno , Sistemas de Mensajero Secundario/fisiología , Propiedades de Superficie
3.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35145026

RESUMEN

Bacteroides thetaiotaomicron is a gut symbiont that inhabits the mucus layer and adheres to and metabolizes food particles, contributing to gut physiology and maturation. Although adhesion and biofilm formation could be key features for B. thetaiotaomicron stress resistance and gut colonization, little is known about the determinants of B. thetaiotaomicron biofilm formation. We previously showed that the B. thetaiotaomicron reference strain VPI-5482 is a poor in vitro biofilm former. Here, we demonstrated that bile, a gut-relevant environmental cue, triggers the formation of biofilm in many B. thetaiotaomicron isolates and common gut Bacteroidales species. We determined that bile-dependent biofilm formation involves the production of the DNase BT3563 or its homologs, degrading extracellular DNA (eDNA) in several B. thetaiotaomicron strains. Our study therefore shows that, although biofilm matrix eDNA provides a biofilm-promoting scaffold in many studied Firmicutes and Proteobacteria, BT3563-mediated eDNA degradation is required to form B. thetaiotaomicron biofilm in the presence of bile.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacteroides thetaiotaomicron/enzimología , Bilis/metabolismo , Biopelículas/crecimiento & desarrollo , Desoxirribonucleasas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Bacterianas/genética , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/fisiología , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Desoxirribonucleasas/genética , Regulación Enzimológica de la Expresión Génica/fisiología
4.
J Biol Chem ; 299(6): 104777, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37142222

RESUMEN

Mycobacterium tuberculosis (Mtb) WhiB3 is an iron-sulfur cluster-containing transcription factor belonging to a subclass of the WhiB-Like (Wbl) family that is widely distributed in the phylum Actinobacteria. WhiB3 plays a crucial role in the survival and pathogenesis of Mtb. It binds to the conserved region 4 of the principal sigma factor (σA4) in the RNA polymerase holoenzyme to regulate gene expression like other known Wbl proteins in Mtb. However, the structural basis of how WhiB3 coordinates with σA4 to bind DNA and regulate transcription is unclear. Here we determined crystal structures of the WhiB3:σA4 complex without and with DNA at 1.5 Å and 2.45 Å, respectively, to elucidate how WhiB3 interacts with DNA to regulate gene expression. These structures reveal that the WhiB3:σA4 complex shares a molecular interface similar to other structurally characterized Wbl proteins and also possesses a subclass-specific Arg-rich DNA-binding motif. We demonstrate that this newly defined Arg-rich motif is required for WhiB3 binding to DNA in vitro and transcriptional regulation in Mycobacterium smegmatis. Together, our study provides empirical evidence of how WhiB3 regulates gene expression in Mtb by partnering with σA4 and engaging with DNA via the subclass-specific structural motif, distinct from the modes of DNA interaction by WhiB1 and WhiB7.


Asunto(s)
Proteínas Bacterianas , Modelos Moleculares , Mycobacterium tuberculosis , Factores de Transcripción , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Estructura Cuaternaria de Proteína , Factor sigma/química , Factor sigma/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo
5.
PLoS Pathog ; 18(1): e1010170, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34986198

RESUMEN

Pseudomonas aeruginosa is an important opportunistic pathogen capable of causing variety of infections in humans. The type III secretion system (T3SS) is a critical virulence determinant of P. aeruginosa in the host infections. Expression of the T3SS is regulated by ExsA, a master regulator that activates the expression of all known T3SS genes. Expression of the exsA gene is controlled at both transcriptional and posttranscriptional levels. Here, we screened a P. aeruginosa transposon (Tn5) insertional mutant library and found rplI, a gene coding for the ribosomal large subunit protein L9, to be a repressor for the T3SS gene expression. Combining real-time quantitative PCR (qPCR), western blotting and lacZ fusion assays, we show that RplI controls the expression of exsA at the posttranscriptional level. Further genetic experiments demonstrated that RplI mediated control of the exsA translation involves 5' untranslated region (5' UTR). A ribosome immunoprecipitation assay and qPCR revealed higher amounts of a 24 nt fragment from exsA mRNA being associated with ribosomes in the ΔrplI mutant. An interaction between RplI and exsA mRNA harboring its 24 nt, but not 12 nt, 5' UTR was confirmed by RNA Gel Mobility Shift and Microscale Thermophoresis assays. Overall, this study identifies the ribosomal large subunit protein L9 as a novel T3SS repressor that inhibits ExsA translation in P. aeruginosa.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Pseudomonas aeruginosa/patogenicidad , Proteínas Ribosómicas/metabolismo , Transactivadores/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Regiones no Traducidas 5' , Células HeLa , Humanos , Pseudomonas aeruginosa/metabolismo , Transcripción Genética , Virulencia/fisiología , Factores de Virulencia/metabolismo
6.
Nat Chem Biol ; 18(2): 161-170, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34931064

RESUMEN

Multi-enzyme assemblies composed of metabolic enzymes catalyzing sequential reactions are being increasingly studied. Here, we report the discovery of a 1.6 megadalton multi-enzyme complex from Bacillus subtilis composed of two enzymes catalyzing opposite ('counter-enzymes') rather than sequential reactions: glutamate synthase (GltAB) and glutamate dehydrogenase (GudB), which make and break glutamate, respectively. In vivo and in vitro studies show that the primary role of complex formation is to inhibit the activity of GudB. Using cryo-electron microscopy, we elucidated the structure of the complex and the molecular basis of inhibition of GudB by GltAB. The complex exhibits unusual oscillatory progress curves and is necessary for both planktonic growth, in glutamate-limiting conditions, and for biofilm growth, in glutamate-rich media. The regulation of a key metabolic enzyme by complexing with its counter enzyme may thus enable cell growth under fluctuating glutamate concentrations.


Asunto(s)
Bacillus subtilis/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Glutamato Deshidrogenasa/metabolismo , Glutamato Sintasa/metabolismo , Ácido Glutámico/biosíntesis , Bacillus subtilis/genética , Proteínas Bacterianas , Glutamato Deshidrogenasa/genética , Glutamato Sintasa/genética
7.
Nat Chem Biol ; 18(2): 142-151, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34903851

RESUMEN

Protein lysine 2-hydroxyisobutyrylation (Khib) has recently been shown to play a critical role in the regulation of cellular processes. However, the mechanism and functional consequence of Khib in prokaryotes remain unclear. Here we report that TmcA, an RNA acetyltransferase, functions as a lysine 2-hydroxyisobutyryltransferase in the regulation of transcription. We show that TmcA can effectively catalyze Khib both in vitro and intracellularly, and that R502 is a key site for the Khib catalytic activity of TmcA. Using quantitative proteomics, we identified 467 endogenous candidates targeted by TmcA for Khib in Escherichia coli. Interestingly, we demonstrate that TmcA can specifically modulate the DNA-binding activity of H-NS, a nucleoid-associated protein, by catalysis of Khib at K121. Furthermore, this TmcA-targeted Khib regulates transcription of acid-resistance genes and enhances E. coli survival under acid stress. Our study reveals transcription regulation mediated by TmcA-catalyzed Khib for bacterial acid resistance.


Asunto(s)
Acetiltransferasas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Acetiltransferasas/genética , Ácidos , Secuencia de Aminoácidos , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Estrés Fisiológico , Transcripción Genética , Transcriptoma
8.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34326261

RESUMEN

Streptomyces genomes harbor numerous, biosynthetic gene clusters (BGCs) encoding for drug-like compounds. While some of these BGCs readily yield expected products, many do not. Biosynthetic crypticity represents a significant hurdle to drug discovery, and the biological mechanisms that underpin it remain poorly understood. Polycyclic tetramate macrolactam (PTM) antibiotic production is widespread within the Streptomyces genus, and examples of active and cryptic PTM BGCs are known. To reveal further insights into the causes of biosynthetic crypticity, we employed a PTM-targeted comparative metabologenomics approach to analyze a panel of S. griseus clade strains that included both poor and robust PTM producers. By comparing the genomes and PTM production profiles of these strains, we systematically mapped the PTM promoter architecture within the group, revealed that these promoters are directly activated via the global regulator AdpA, and discovered that small promoter insertion-deletion lesions (indels) differentiate weaker PTM producers from stronger ones. We also revealed an unexpected link between robust PTM expression and griseorhodin pigment coproduction, with weaker S. griseus-clade PTM producers being unable to produce the latter compound. This study highlights promoter indels and biosynthetic interactions as important, genetically encoded factors that impact BGC outputs, providing mechanistic insights that will undoubtedly extend to other Streptomyces BGCs. We highlight comparative metabologenomics as a powerful approach to expose genomic features that differentiate strong, antibiotic producers from weaker ones. This should prove useful for rational discovery efforts and is orthogonal to current engineering and molecular signaling approaches now standard in the field.


Asunto(s)
Antibacterianos/metabolismo , Genómica , Streptomyces/metabolismo , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Genoma Bacteriano , Regiones Promotoras Genéticas , Streptomyces/genética
9.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34907018

RESUMEN

Photosynthetic reaction centers (RCs) from Rhodobacter sphaeroides were engineered to vary the electronic properties of a key tyrosine (M210) close to an essential electron transfer component via its replacement with site-specific, genetically encoded noncanonical amino acid tyrosine analogs. High fidelity of noncanonical amino acid incorporation was verified with mass spectrometry and X-ray crystallography and demonstrated that RC variants exhibit no significant structural alterations relative to wild type (WT). Ultrafast transient absorption spectroscopy indicates the excited primary electron donor, P*, decays via a ∼4-ps and a ∼20-ps population to produce the charge-separated state P+HA- in all variants. Global analysis indicates that in the ∼4-ps population, P+HA- forms through a two-step process, P*→ P+BA-→ P+HA-, while in the ∼20-ps population, it forms via a one-step P* → P+HA- superexchange mechanism. The percentage of the P* population that decays via the superexchange route varies from ∼25 to ∼45% among variants, while in WT, this percentage is ∼15%. Increases in the P* population that decays via superexchange correlate with increases in the free energy of the P+BA- intermediate caused by a given M210 tyrosine analog. This was experimentally estimated through resonance Stark spectroscopy, redox titrations, and near-infrared absorption measurements. As the most energetically perturbative variant, 3-nitrotyrosine at M210 creates an ∼110-meV increase in the free energy of P+BA- along with a dramatic diminution of the 1,030-nm transient absorption band indicative of P+BA- formation. Collectively, this work indicates the tyrosine at M210 tunes the mechanism of primary electron transfer in the RC.


Asunto(s)
Proteínas Bacterianas/metabolismo , Variación Genética , Proteínas del Complejo del Centro de Reacción Fotosintética/genética , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/fisiología , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Transporte de Electrón , Regulación Bacteriana de la Expresión Génica/fisiología , Conformación Proteica
10.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34385314

RESUMEN

Structural maintenance of chromosomes (SMC) complexes contribute to chromosome organization in all domains of life. In Escherichia coli, MukBEF, the functional SMC homolog, promotes spatiotemporal chromosome organization and faithful chromosome segregation. Here, we address the relative contributions of MukBEF and the replication terminus (ter) binding protein, MatP, to chromosome organization-segregation. We show that MukBEF, but not MatP, is required for the normal localization of the origin of replication to midcell and for the establishment of translational symmetry between newly replicated sister chromosomes. Overall, chromosome orientation is normally maintained through division from one generation to the next. Analysis of loci flanking the replication termination region (ter), which demark the ends of the linearly organized portion of the nucleoid, demonstrates that MatP is required for maintenance of chromosome orientation. We show that DNA-bound ß2-processivity clamps, which mark the lagging strands at DNA replication forks, localize to the cell center, independent of replisome location but dependent on MukBEF action, and consistent with translational symmetry of sister chromosomes. Finally, we directly show that the older ("immortal") template DNA strand, propagated from previous generations, is preferentially inherited by the cell forming at the old pole, dependent on MukBEF and MatP. The work further implicates MukBEF and MatP as central players in chromosome organization, segregation, and nonrandom inheritance of genetic material and suggests a general framework for understanding how chromosome conformation and dynamics shape subcellular organization.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica/fisiología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/fisiología , Proteínas Represoras/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas de Escherichia coli/genética , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología
11.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34389683

RESUMEN

Recently discovered simple quantitative relations, known as bacterial growth laws, hint at the existence of simple underlying principles at the heart of bacterial growth. In this work, we provide a unifying picture of how these known relations, as well as relations that we derive, stem from a universal autocatalytic network common to all bacteria, facilitating balanced exponential growth of individual cells. We show that the core of the cellular autocatalytic network is the transcription-translation machinery-in itself an autocatalytic network comprising several coupled autocatalytic cycles, including the ribosome, RNA polymerase, and transfer RNA (tRNA) charging cycles. We derive two types of growth laws per autocatalytic cycle, one relating growth rate to the relative fraction of the catalyst and its catalysis rate and the other relating growth rate to all the time scales in the cycle. The structure of the autocatalytic network generates numerous regimes in state space, determined by the limiting components, while the number of growth laws can be much smaller. We also derive a growth law that accounts for the RNA polymerase autocatalytic cycle, which we use to explain how growth rate depends on the inducible expression of the rpoB and rpoC genes, which code for the RpoB and C protein subunits of RNA polymerase, and how the concentration of rifampicin, which targets RNA polymerase, affects growth rate without changing the RNA-to-protein ratio. We derive growth laws for tRNA synthesis and charging and predict how growth rate depends on temperature, perturbation to ribosome assembly, and membrane synthesis.


Asunto(s)
Bacterias/metabolismo , Proliferación Celular/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , ARN Bacteriano/metabolismo , Bacterias/genética , Fenómenos Fisiológicos Bacterianos , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Modelos Biológicos , ARN Bacteriano/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Ribosomas/fisiología , Transcripción Genética
12.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34873041

RESUMEN

The treatment of infectious diseases caused by multidrug-resistant pathogens is a major clinical challenge of the 21st century. The membrane-embedded respiratory cytochrome bd-type oxygen reductase is a critical survival factor utilized by pathogenic bacteria during infection, proliferation and the transition from acute to chronic states. Escherichia coli encodes for two cytochrome bd isoforms that are both involved in respiration under oxygen limited conditions. Mechanistic and structural differences between cydABX (Ecbd-I) and appCBX (Ecbd-II) operon encoded cytochrome bd variants have remained elusive in the past. Here, we demonstrate that cytochrome bd-II catalyzes oxidation of benzoquinols while possessing additional specificity for naphthoquinones. Our data show that although menaquinol-1 (MK1) is not able to directly transfer electrons onto cytochrome bd-II from E. coli, it has a stimulatory effect on its oxygen reduction rate in the presence of ubiquinol-1. We further determined cryo-EM structures of cytochrome bd-II to high resolution of 2.1 Å. Our structural insights confirm that the general architecture and substrate accessible pathways are conserved between the two bd oxidase isoforms, but two notable differences are apparent upon inspection: (i) Ecbd-II does not contain a CydH-like subunit, thereby exposing heme b595 to the membrane environment and (ii) the AppB subunit harbors a structural demethylmenaquinone-8 molecule instead of ubiquinone-8 as found in CydB of Ecbd-I Our work completes the structural landscape of terminal respiratory oxygen reductases of E. coli and suggests that structural and functional properties of the respective oxidases are linked to quinol-pool dependent metabolic adaptations in E. coli.


Asunto(s)
Grupo Citocromo b/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Oxidorreductasas/metabolismo , Grupo Citocromo b/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Modelos Moleculares , Oxidorreductasas/genética , Conformación Proteica , Isoformas de Proteínas
13.
PLoS Pathog ; 17(3): e1009375, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33690714

RESUMEN

Pseudomonas aeruginosa causes chronic airway infections, a major determinant of lung inflammation and damage in cystic fibrosis (CF). Loss-of-function lasR mutants commonly arise during chronic CF infections, are associated with accelerated lung function decline in CF patients and induce exaggerated neutrophilic inflammation in model systems. In this study, we investigated how lasR mutants modulate airway epithelial membrane bound ICAM-1 (mICAM-1), a surface adhesion molecule, and determined its impact on neutrophilic inflammation in vitro and in vivo. We demonstrated that LasR-deficient strains induce increased mICAM-1 levels in airway epithelial cells compared to wild-type strains, an effect attributable to the loss of mICAM-1 degradation by LasR-regulated proteases and associated with enhanced neutrophil adhesion. In a subacute airway infection model, we also observed that lasR mutant-infected mice displayed greater airway epithelial ICAM-1 expression and increased neutrophilic pulmonary inflammation. Our findings provide new insights into the intricate interplay between lasR mutants, LasR-regulated proteases and airway epithelial ICAM-1 expression, and reveal a new mechanism involved in the exaggerated inflammatory response induced by lasR mutants.


Asunto(s)
Fibrosis Quística/complicaciones , Neumonía/microbiología , Pseudomonas aeruginosa/patogenicidad , Sistema Respiratorio/parasitología , Animales , Proteínas Bacterianas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Humanos , Ratones , Neumonía/complicaciones , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa/metabolismo , Sistema Respiratorio/metabolismo , Transactivadores/genética
14.
Nat Chem Biol ; 17(10): 1016-1026, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34552219

RESUMEN

Escherichia coli are a common component of the human microbiota, and isolates exhibit probiotic, commensal and pathogenic roles in the host. E. coli members often use diverse small molecule chemistry to regulate intrabacterial, intermicrobial and host-bacterial interactions. While E. coli are considered to be a well-studied model organism in biology, much of their chemical arsenal has only more recently been defined, and much remains to be explored. Here we describe chemical signaling systems in E. coli in the context of the broader field of metabolism at the host-bacteria interface and the role of this signaling in disease modulation.


Asunto(s)
Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Interacciones Microbiota-Huesped/fisiología , Humanos , Microbiota , Probióticos
15.
Nat Chem Biol ; 17(10): 1093-1100, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34326538

RESUMEN

Gram-negative bacteria express structurally diverse lipoproteins in their cell envelope. Here, we find that approximately half of lipoproteins destined to the Escherichia coli outer membrane display an intrinsically disordered linker at their N terminus. Intrinsically disordered regions are common in proteins, but establishing their importance in vivo has remained challenging. As we sought to unravel how lipoproteins mature, we discovered that unstructured linkers are required for optimal trafficking by the Lol lipoprotein sorting system, whereby linker deletion re-routes three unrelated lipoproteins to the inner membrane. Focusing on the stress sensor RcsF, we found that replacing the linker with an artificial peptide restored normal outer-membrane targeting only when the peptide was of similar length and disordered. Overall, this study reveals the role played by intrinsic disorder in lipoprotein sorting, providing mechanistic insight into the biogenesis of these proteins and suggesting that evolution can select for intrinsic disorder that supports protein function.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Lipoproteínas/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Intrínsecamente Desordenadas/química , Lipoproteínas/genética , Modelos Moleculares , Conformación Proteica , Transporte de Proteínas
16.
PLoS Biol ; 18(12): e3000952, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33270631

RESUMEN

Populations of bacteria often undergo a lag in growth when switching conditions. Because growth lags can be large compared to typical doubling times, variations in growth lag are an important but often overlooked component of bacterial fitness in fluctuating environments. We here explore how growth lag variation is determined for the archetypical switch from glucose to lactose as a carbon source in Escherichia coli. First, we show that single-cell lags are bimodally distributed and controlled by a single-molecule trigger. That is, gene expression noise causes the population before the switch to divide into subpopulations with zero and nonzero lac operon expression. While "sensorless" cells with zero preexisting lac expression at the switch have long lags because they are unable to sense the lactose signal, any nonzero lac operon expression suffices to ensure a short lag. Second, we show that the growth lag at the population level depends crucially on the fraction of sensorless cells and that this fraction in turn depends sensitively on the growth condition before the switch. Consequently, even small changes in basal expression can significantly affect the fraction of sensorless cells, thereby population lags and fitness under switching conditions, and may thus be subject to significant natural selection. Indeed, we show that condition-dependent population lags vary across wild E. coli isolates. Since many sensory genes are naturally low expressed in conditions where their inducer is not present, bimodal responses due to subpopulations of sensorless cells may be a general mechanism inducing phenotypic heterogeneity and controlling population lags in switching environments. This mechanism also illustrates how gene expression noise can turn even a simple sensory gene circuit into a bet hedging module and underlines the profound role of gene expression noise in regulatory responses.


Asunto(s)
Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Aptitud Genética/fisiología , Bacterias/genética , Bacterias/metabolismo , Ambiente , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Redes Reguladoras de Genes/genética , Interacción Gen-Ambiente , Aptitud Genética/genética , Glucosa/metabolismo , Operón Lac , Lactosa/metabolismo , Fenotipo
17.
Fish Shellfish Immunol ; 134: 108639, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36841518

RESUMEN

High temperature is a main cause to result in the outbreak of tilapia streptococcal disease. However, the underlying mechanisms are not well understood. In this study, we first confirmed that tilapia infected with Streptococcus agalactiae (S. agalactiae) had a higher mortality at high temperature (35 °C) than that at normal temperature (28 °C). Subsequently, the effects of high temperature on gene expression pattern of S. agalactiae and intestinal microbiota of tilapia were respectively detected by RNA-seq and 16S rDNA sequencing. RNA-seq identified 357 differentially expressed genes (DEGs) in S. agalactiae cultured at 28 °C and 35 °C. GO and KEGG analysis showed that these DEGs were highly involved in metabolic processes, including glucose, lipid and amino acid metabolisms, which indicates that S. agalactiae have stronger vitality and are likely to be more infectious under high temperature. Microbiota analysis revealed that high temperature could influence the bacterial community composition of tilapia intestine, accompanied by changes in intestinal structure. Compared to feed at 28 °C, the total bacterial species as well as pathogens, such as norank_f__Rhizobiales_Incertae_Sedis, Pseudorhodoplanes, Ancylobacter, in tilapia intestine were significantly increased at 35 °C, which may weaken the immune resistance of tilapia. Taken together, our results suggest that high temperature evoked tilapia susceptible to S. agalactiae should be the combined effect of enhanced S. agalactiae metabolism and dysregulated tilapia intestinal microbiota.


Asunto(s)
Brotes de Enfermedades , Enfermedades de los Peces , Regulación Bacteriana de la Expresión Génica , Calor , Infecciones Estreptocócicas , Streptococcus agalactiae , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/microbiología , Infecciones Estreptocócicas/veterinaria , Enfermedades de los Peces/epidemiología , Enfermedades de los Peces/microbiología , Tilapia , Microbioma Gastrointestinal/fisiología , Regulación Bacteriana de la Expresión Génica/fisiología , Biodiversidad , Animales
18.
Proc Natl Acad Sci U S A ; 117(17): 9546-9553, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32295877

RESUMEN

Understanding how bacteria colonize surfaces and regulate cell-cycle progression in response to cellular adhesion is of fundamental importance. Here, we use transposon sequencing in conjunction with fluorescence resonance energy transfer (FRET) microscopy to uncover the molecular mechanism for how surface sensing drives cell-cycle initiation in Caulobacter crescentus We identify the type IV pilin protein PilA as the primary signaling input that couples surface contact to cell-cycle initiation via the second messenger cyclic di-GMP (c-di-GMP). Upon retraction of pili filaments, the monomeric pilin reservoir in the inner membrane is sensed by the 17-amino acid transmembrane helix of PilA to activate the PleC-PleD two-component signaling system, increase cellular c-di-GMP levels, and signal the onset of the cell cycle. We termed the PilA signaling sequence CIP for "cell-cycle initiating pilin" peptide. Addition of the chemically synthesized CIP peptide initiates cell-cycle progression and simultaneously inhibits surface attachment. The broad conservation of the type IV pili and their importance in pathogens for host colonization suggests that CIP peptide mimetics offer strategies to inhibit surface sensing, prevent biofilm formation and control persistent infections.


Asunto(s)
Adhesión Bacteriana/fisiología , Caulobacter crescentus/fisiología , Ciclo Celular/fisiología , Proteínas Fimbrias/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Fimbrias/genética
19.
Proc Natl Acad Sci U S A ; 117(17): 9519-9528, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32277032

RESUMEN

Enterohemorrhagic Escherichia coli (EHEC) is a foodborne pathogen that colonizes the gastrointestinal tract and has evolved intricate mechanisms to sense and respond to the host environment. Upon the sensation of chemical and physical cues specific to the host's intestinal environment, locus of enterocyte effacement (LEE)-encoded virulence genes are activated and promote intestinal colonization. The LEE transcriptional activator GrlA mediates EHEC's response to mechanical cues characteristic of the intestinal niche, including adhesive force that results from bacterial adherence to epithelial cells and fluid shear that results from intestinal motility and transit. GrlA expression and release from its inhibitor GrlR was not sufficient to induce virulence gene transcription; mechanical stimuli were required for GrlA activation. The exact mechanism of GrlA activation, however, remained unknown. We isolated GrlA mutants that activate LEE transcription, independent of applied mechanical stimuli. In nonstimulated EHEC, wild-type GrlA associates with cardiolipin membrane domains via a patch of basic C-terminal residues, and this membrane sequestration is disrupted in EHEC that expresses constitutively active GrlA mutants. GrlA transitions from an inactive, membrane-associated state and relocalizes to the cytoplasm in response to mechanical stimuli, allowing GrlA to bind and activate the LEE1 promoter. GrlA expression and its relocalization in response to mechanical stimuli are required for optimal virulence regulation and colonization of the host intestinal tract during infection. These data suggest a posttranslational regulatory mechanism of the mechanosensor GrlA, whereby virulence gene expression can be rapidly fine-tuned in response to the highly dynamic spatiotemporal mechanical profile of the gastrointestinal tract.


Asunto(s)
Escherichia coli O157/metabolismo , Proteínas de Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Transactivadores/metabolismo , Animales , Proteínas de Escherichia coli/genética , Larva/microbiología , Mecanotransducción Celular , Mutación Puntual , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Transactivadores/genética , Transcripción Genética , Virulencia , Pez Cebra
20.
Proc Natl Acad Sci U S A ; 117(36): 22167-22172, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32839318

RESUMEN

Accurate protein synthesis is a tightly controlled biological process with multiple quality control steps safeguarded by aminoacyl-transfer RNA (tRNA) synthetases and the ribosome. Reduced translational accuracy leads to various physiological changes in both prokaryotes and eukaryotes. Termination of translation is signaled by stop codons and catalyzed by release factors. Occasionally, stop codons can be suppressed by near-cognate aminoacyl-tRNAs, resulting in protein variants with extended C termini. We have recently shown that stop-codon readthrough is heterogeneous among single bacterial cells. However, little is known about how environmental factors affect the level and heterogeneity of stop-codon readthrough. In this study, we have combined dual-fluorescence reporters, mass spectrometry, mathematical modeling, and single-cell approaches to demonstrate that a metabolic stress caused by excess carbon substantially increases both the level and heterogeneity of stop-codon readthrough. Excess carbon leads to accumulation of acid metabolites, which lower the pH and the activity of release factors to promote readthrough. Furthermore, our time-lapse microscopy experiments show that single cells with high readthrough levels are more adapted to severe acid stress conditions and are more sensitive to an aminoglycoside antibiotic. Our work thus reveals a metabolic stress that promotes translational heterogeneity and phenotypic diversity.


Asunto(s)
Codón de Terminación , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Concentración de Iones de Hidrógeno , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA